Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Anal Chem ; 94(28): 10045-10053, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35792073

RESUMO

The phosphonate group is a key pharmacophore in many antiviral, antimicrobial, and antineoplastic drugs. Due to its high polarity and short retention time, detecting and quantifying such phosphonate-containing drugs with LC/MS-based methods are challenging and require derivatization with hazardous reagents. Given the emerging importance of phosphonate-containing drugs, developing a practical, accessible, and safe method for their quantitation in pharmacokinetics (PK) studies is desirable. NMR-based methods are often employed in drug discovery but are seldom used for compound quantitation in PK studies. Here, we show that proton-phosphorous (1H-31P) heteronuclear single quantum correlation (HSQC) NMR allows for the quantitation of the phosphonate-containing enolase inhibitor HEX in plasma and tissues at micromolar concentrations. Although mice were shown to rapidly clear HEX from circulation (over 95% in <1 h), the plasma half-life of HEX was more than 1 h in rats and nonhuman primates. This slower clearance rate affords a significantly higher exposure of HEX in rat models compared to that in mouse models while maintaining a favorable safety profile. Similar results were observed for the phosphonate-containing antibiotic, fosfomycin. Our study demonstrates the applicability of the 1H-31P HSQC method to quantify phosphonate-containing drugs in complex biological samples and illustrates an important limitation of mice as preclinical model species for phosphonate-containing drugs.


Assuntos
Antineoplásicos , Organofosfonatos , Animais , Antineoplásicos/farmacocinética , Antivirais , Camundongos , Organofosfonatos/química , Primatas , Prótons , Ratos
2.
Antimicrob Agents Chemother ; 59(8): 4889-900, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26055364

RESUMO

Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in infants. Effective treatment for RSV infection is a significant unmet medical need. While new RSV therapeutics are now in development, there are very few animal models that mimic the pathogenesis of human RSV, making it difficult to evaluate new disease interventions. Experimental infection of Holstein calves with bovine RSV (bRSV) causes a severe respiratory infection that is similar to human RSV infection, providing a relevant model for testing novel therapeutic agents. In this model, viral load is readily detected in nasal secretions by quantitative real-time PCR (qRT-PCR), and cumulative symptom scoring together with histopathology evaluations of infected tissue allow for the assessment of disease severity. The bovine RSV model was used to evaluate the antiviral activity of an RSV fusion inhibitor, GS1, which blocks virus entry by inhibiting the fusion of the viral envelope with the host cell membrane. The efficacy of GS1, a close structural analog of GS-5806 that is being developed to treat RSV infection in humans was evaluated in two randomized, blind, placebo-controlled studies in bRSV-infected calves. Intravenous administration of GS1 at 4 mg/kg of body weight/day for 7 days starting 24 h or 72 h postinoculation provided clear therapeutic benefit by reducing the viral load, disease symptom score, respiration rate, and lung pathology associated with bRSV infection. These data support the use of the bovine RSV model for evaluation of experimental therapeutics for treatment of RSV.


Assuntos
Antivirais/farmacologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Bovino/efeitos dos fármacos , Animais , Bronquiolite/tratamento farmacológico , Bronquiolite/virologia , Bovinos , Doenças dos Bovinos/tratamento farmacológico , Doenças dos Bovinos/patologia , Doenças dos Bovinos/virologia , Linhagem Celular , Membrana Celular/patologia , Membrana Celular/virologia , Método Duplo-Cego , Humanos , Indazóis , Pulmão/patologia , Pulmão/virologia , Masculino , Pirazóis/farmacologia , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Sulfonamidas/farmacologia , Carga Viral/efeitos dos fármacos
3.
J Clin Pharmacol ; 58(6): 771-780, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29412463

RESUMO

Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in young children. Presatovir (previously GS-5806) is a novel, orally administered RSV fusion inhibitor with a favorable safety profile and proven antiviral efficacy in preclinical and clinical studies. In vitro, presatovir is a substrate of the efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) and hepatic uptake transporters organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 and is slowly metabolized by cytochrome P450 (CYP) 3A4 and CYP3A5. This study enrolled 64 healthy subjects to evaluate the effect of cyclosporine, a P-gp, BCRP, and OATP1B1/1B3 inhibitor; rifampin, a strong CYP3A4 and P-gp inducer; efavirenz, a moderate CYP3A4 inducer; and cobicistat, a potent CYP3A inhibitor, on presatovir pharmacokinetics. Presatovir plasma exposures (maximum observed plasma concentration [Cmax ] and area under the plasma concentration-time curve from time 0 extrapolated to infinity [AUCinf ]) were not affected by coadministration of cyclosporine, suggesting presatovir is not a sensitive substrate of P-gp, BCRP, or OATP1B1/1B3. As expected, based on the role of CYP3A in presatovir metabolism, presatovir exposure was increased by cobicistat (122% in AUCinf ), and decreased by rifampin (40.3% in Cmax and 82.5% in AUCinf ) and efavirenz (55.7% in AUCinf ). These data support coadministration of presatovir with inhibitors of P-gp, BCRP, OATP1B1/1B3, or CYP3A, but not with moderate or strong CYP3A4 inducers. Presatovir was well-tolerated with the most common drug-related adverse events of dizziness (n = 12) and somnolence (n = 4) reported during efavirenz treatment.


Assuntos
Antivirais/farmacologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Adolescente , Adulto , Alcinos , Antivirais/efeitos adversos , Antivirais/sangue , Antivirais/farmacocinética , Área Sob a Curva , Benzoxazinas/farmacocinética , Cobicistat/farmacocinética , Ciclopropanos , Ciclosporina/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Humanos , Indazóis , Masculino , Pessoa de Meia-Idade , Pirazóis/efeitos adversos , Pirazóis/sangue , Pirazóis/farmacocinética , Rifampina/farmacocinética , Sulfonamidas/efeitos adversos , Sulfonamidas/sangue , Sulfonamidas/farmacocinética
4.
Antivir Ther ; 10(3): 451-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15918336

RESUMO

OBJECTIVE: An anti-HIV regimen composed of the nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) tenofovir (TFV) disoproxil fumarate (TDF), abacavir (ABC) and lamivudine (3TC) has performed poorly in patients. This study evaluated the combination of TFV, ABC and 3TC for metabolic or antiviral antagonism in vitro. DESIGN: Procedures were developed to evaluate the in vitro metabolism and antiviral activity of drug combinations of TFV, ABC and 3TC in cell types relevant for HIV infection. METHODS: Anabolism of combinations of TFV and ABC were studied over a 24 h period in the human T leukaemic CEM lymphoblast cell line and human primary peripheral blood mononuclear cells (PBMCs) stimulated with human interleukin-2 and phytohaemagglutinin. The anti-HIV activity of combinations of TFV and ABC in the presence or absence of 3TC was studied in stimulated PBMCs infected with the HXB2 strain of HIV-1. RESULTS: Levels of the active metabolites produced from TFV and ABC after incubation with CEM or PBMCs showed no significant change upon introduction of the other NRTI. Moreover, the pool sizes for the natural substrates of 2'-deoxyadenosine triphosphate and 2'-deoxyguanosine triphosphate were also unchanged. In anti-HIV assays in PBMCs, the combination of TFV and ABC was found to be additive with respect to inhibition of HIV replication. Addition of 3TC to the combination did not result in synergistic or antagonistic effects. CONCLUSIONS: The poor efficacy of the triple NRTI regimen of TDF, ABC and 3TC is probably not due to a metabolic drug interaction resulting in antagonism of antiviral activity.


Assuntos
Adenina/análogos & derivados , Adenina/farmacologia , Fármacos Anti-HIV/farmacologia , Didesoxinucleosídeos/farmacologia , HIV-1/efeitos dos fármacos , Lamivudina/farmacologia , Organofosfonatos/farmacologia , Adenina/metabolismo , Fármacos Anti-HIV/metabolismo , Linhagem Celular , Didesoxinucleosídeos/metabolismo , Interações Medicamentosas , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Organofosfonatos/metabolismo , Tenofovir
5.
ACS Med Chem Lett ; 2(10): 715-9, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900257

RESUMO

A novel series of HCV replication inhibitors based on a pyrido[3,2-d]pyrimidine core were optimized for pharmacokinetics (PK) in rats. Several associations between physicochemical properties and PK were identified and exploited to guide the design of compounds. In addition, a simple new metric that may aid in the prediction of bioavailability for compounds with higher polar surface area is described (3*HBD-cLogP).

6.
J Med Chem ; 52(19): 5781-4, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19791799

RESUMO

Pyrimidinol carboxylic acids were designed as inhibitors of HIV-1 RNase H function. These molecules can coordinate to two divalent metal ions in the RNase H active site. Inhibition of enzymatic activity was measured in a biochemical assay, but no antiviral effect was observed. Binding was demonstrated via a solid state structure of the isolated p15-Ec domain of HIV-1 RT showing inhibitor and two Mn(II) ions bound to the RNase H active site.


Assuntos
Transcriptase Reversa do HIV/antagonistas & inibidores , Pirimidinas/farmacologia , Ribonuclease H/antagonistas & inibidores , Ácidos Carboxílicos , Domínio Catalítico , Desenho de Fármacos , Humanos , Ligação Proteica , Pirimidinas/química
7.
Antimicrob Agents Chemother ; 50(10): 3297-304, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17005808

RESUMO

Tenofovir (TFV) undergoes renal elimination by a combination of glomerular filtration and active tubular secretion. While transporter-mediated uptake of TFV from the blood into proximal-tubule cells has been well characterized, comparatively little is known about the efflux system responsible for transporting TFV into the lumen during active tubular secretion. Therefore, members of the ATP-binding cassette family of efflux pumps expressed at the apical side of proximal-tubule cells were studied for the ability to transport TFV. Studies in multiple independent in vitro systems show TFV not to be a substrate for P glycoprotein (Pgp) or multidrug resistance protein type 2 (MRP2). In contrast to Pgp and MRP2, TFV was observed to be a substrate for MRP4. TFV accumulated to fivefold lower levels in MRP4-overexpressing cells, and its accumulation could be increased by an MRP inhibitor. Furthermore, MRP4-overexpressing cells were found to be 2.0- to 2.5-fold less susceptible to cytotoxicity caused by TFV. ATP-dependent uptake of TFV was observed in membrane vesicles containing MRP4 but not in vesicles lacking the transporter. On the basis of these and previous results, the molecular transport pathway for the active tubular secretion of TFV through renal proximal-tubule cells involves uptake from the blood mediated by human organic anion transporters 1 and 3 and efflux into urine by MRP4. A detailed understanding of the molecular mechanism of TFV active tubular secretion will facilitate the assessment of potential renal drug-drug interactions with coadministered agents.


Assuntos
Adenina/análogos & derivados , Fármacos Anti-HIV/farmacocinética , Túbulos Renais Proximais/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Organofosfonatos/farmacocinética , Inibidores da Transcriptase Reversa/farmacocinética , Adenina/farmacocinética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Cães , Humanos , Transportadores de Ânions Orgânicos , Tenofovir
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa